Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 709
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 176(1-2): 334-347.e12, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30580966

RESUMEN

Lymphocyte-activation gene 3 (LAG-3) is an immune inhibitory receptor, with major histocompatibility complex class II (MHC-II) as a canonical ligand. However, it remains controversial whether MHC-II is solely responsible for the inhibitory function of LAG-3. Here, we demonstrate that fibrinogen-like protein 1 (FGL1), a liver-secreted protein, is a major LAG-3 functional ligand independent from MHC-II. FGL1 inhibits antigen-specific T cell activation, and ablation of FGL1 in mice promotes T cell immunity. Blockade of the FGL1-LAG-3 interaction by monoclonal antibodies stimulates tumor immunity and is therapeutic against established mouse tumors in a receptor-ligand inter-dependent manner. FGL1 is highly produced by human cancer cells, and elevated FGL1 in the plasma of cancer patients is associated with a poor prognosis and resistance to anti-PD-1/B7-H1 therapy. Our findings reveal an immune evasion mechanism and have implications for the design of cancer immunotherapy.


Asunto(s)
Antígenos CD/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/fisiología , Animales , Antígenos CD/inmunología , Línea Celular , Fibrinógeno/inmunología , Fibrinógeno/metabolismo , Genes MHC Clase II/genética , Genes MHC Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Inmunoterapia , Ligandos , Hígado/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Neoplasias/genética , Neoplasias/inmunología , Linfocitos T Citotóxicos/inmunología , Proteína del Gen 3 de Activación de Linfocitos
2.
Immunity ; 52(1): 136-150.e6, 2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31940267

RESUMEN

Effector CD8+ T cells are important mediators of adaptive immunity, and receptor-ligand interactions that regulate their survival may have therapeutic potential. Here, we identified a subset of effector CD8+ T cells that expressed the inhibitory fragment crystallizable (Fc) receptor FcγRIIB following activation and multiple rounds of division. CD8+ T cell-intrinsic genetic deletion of Fcgr2b increased CD8+ effector T cell accumulation, resulting in accelerated graft rejection and decreased tumor volume in mouse models. Immunoglobulin G (IgG) antibody was not required for FcγRIIB-mediated control of CD8+ T cell immunity, and instead, the immunosuppressive cytokine fibrinogen-like 2 (Fgl2) was a functional ligand for FcγRIIB on CD8+ T cells. Fgl2 induced caspase-3/7-mediated apoptosis in Fcgr2b+, but not Fcgr2b-/-, CD8+ T cells. Increased expression of FcγRIIB correlated with freedom from rejection following withdrawal from immunosuppression in a clinical trial of kidney transplant recipients. Together, these findings demonstrate a cell-intrinsic coinhibitory function of FcγRIIB in regulating CD8+ T cell immunity.


Asunto(s)
Apoptosis/inmunología , Linfocitos T CD8-positivos/inmunología , Fibrinógeno/inmunología , Receptores de IgG/inmunología , Adulto , Anciano , Animales , Caspasa 3/inmunología , Caspasa 7/inmunología , Línea Celular Tumoral , Femenino , Fibrinógeno/genética , Rechazo de Injerto/inmunología , Humanos , Inmunoglobulina G/inmunología , Terapia de Inmunosupresión , Masculino , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Receptores de IgG/genética , Adulto Joven
3.
PLoS Pathog ; 18(1): e1010227, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35041705

RESUMEN

The blood-clotting protein fibrin(ogen) plays a critical role in host defense against invading pathogens, particularly against peritoneal infection by the Gram-positive microbe Staphylococcus aureus. Here, we tested the hypothesis that direct binding between fibrin(ogen) and S. aureus is a component of the primary host antimicrobial response mechanism and prevention of secondary microbe dissemination from the peritoneal cavity. To establish a model system, we showed that fibrinogen isolated from FibγΔ5 mice, which express a mutant form lacking the final 5 amino acids of the fibrinogen γ chain (termed fibrinogenγΔ5), did not support S. aureus adherence when immobilized and clumping when in suspension. In contrast, purified wildtype fibrinogen supported robust adhesion and clumping that was largely dependent on S. aureus expression of the receptor clumping factor A (ClfA). Following peritoneal infection with S. aureus USA300, FibγΔ5 mice displayed worse survival compared to WT mice coupled to reduced bacterial killing within the peritoneal cavity and increased dissemination of the microbes into circulation and distant organs. The failure of acute bacterial killing, but not enhanced dissemination, was partially recapitulated by mice infected with S. aureus USA300 lacking ClfA. Fibrin polymer formation and coagulation transglutaminase Factor XIII each contributed to killing of the microbes within the peritoneal cavity, but only elimination of polymer formation enhanced systemic dissemination. Host macrophage depletion or selective elimination of the fibrin(ogen) ß2-integrin binding motif both compromised local bacterial killing and enhanced S. aureus systemic dissemination, suggesting fibrin polymer formation in and of itself was not sufficient to retain S. aureus within the peritoneal cavity. Collectively, these findings suggest that following peritoneal infection, the binding of S. aureus to stabilized fibrin matrices promotes a local, macrophage-mediated antimicrobial response essential for prevention of microbe dissemination and downstream host mortality.


Asunto(s)
Fibrinógeno/inmunología , Peritonitis/inmunología , Infecciones Estafilocócicas/inmunología , Animales , Coagulasa/inmunología , Coagulasa/metabolismo , Fibrina/metabolismo , Ratones , Peritonitis/metabolismo , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/inmunología , Staphylococcus aureus/metabolismo
4.
Immunity ; 40(4): 569-81, 2014 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-24745333

RESUMEN

Foxp3(+) T regulatory (Treg) cells regulate immune responses and maintain self-tolerance. Recent work shows that Treg cells are comprised of many subpopulations with specialized regulatory functions. Here we identified Foxp3(+) T cells expressing the coinhibitory molecule TIGIT as a distinct Treg cell subset that specifically suppresses proinflammatory T helper 1 (Th1) and Th17 cell, but not Th2 cell responses. Transcriptional profiling characterized TIGIT(+) Treg cells as an activated Treg cell subset with high expression of Treg signature genes. Ligation of TIGIT on Treg cells induced expression of the effector molecule fibrinogen-like protein 2 (Fgl2), which promoted Treg-cell-mediated suppression of T effector cell proliferation. In addition, Fgl2 was necessary to prevent suppression of Th2 cytokine production in a model of allergic airway inflammation. TIGIT expression therefore identifies a Treg cell subset that demonstrates selectivity for suppression of Th1 and Th17 cell but not Th2 cell responses.


Asunto(s)
Fibrinógeno/metabolismo , Receptores Inmunológicos/metabolismo , Hipersensibilidad Respiratoria/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Animales , Proliferación Celular , Células Cultivadas , Citocinas/metabolismo , Eosinófilos/inmunología , Fibrinógeno/genética , Fibrinógeno/inmunología , Factores de Transcripción Forkhead/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Terapia de Inmunosupresión , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Balance Th1 - Th2
5.
J Immunol ; 205(12): 3247-3262, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33168576

RESUMEN

T follicular regulatory (TFR) cells limit Ab responses, but the underlying mechanisms remain largely unknown. In this study, we identify Fgl2 as a soluble TFR cell effector molecule through single-cell gene expression profiling. Highly expressed by TFR cells, Fgl2 directly binds to B cells, especially light-zone germinal center B cells, as well as to T follicular helper (TFH) cells, and directly regulates B cells and TFH in a context-dependent and type 2 Ab isotype-specific manner. In TFH cells, Fgl2 induces the expression of Prdm1 and a panel of checkpoint molecules, including PD1, TIM3, LAG3, and TIGIT, resulting in TFH cell dysfunction. Mice deficient in Fgl2 had dysregulated Ab responses at steady-state and upon immunization. In addition, loss of Fgl2 results in expansion of autoreactive B cells upon immunization. Consistent with this observation, aged Fgl2-/- mice spontaneously developed autoimmunity associated with elevated autoantibodies. Thus, Fgl2 is a TFR cell effector molecule that regulates humoral immunity and limits systemic autoimmunity.


Asunto(s)
Formación de Anticuerpos , Autoanticuerpos/inmunología , Enfermedades Autoinmunes/inmunología , Linfocitos B/inmunología , Fibrinógeno/inmunología , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Enfermedades Autoinmunes/genética , Fibrinógeno/genética , Receptor 2 Celular del Virus de la Hepatitis A/genética , Receptor 2 Celular del Virus de la Hepatitis A/inmunología , Ratones , Ratones Noqueados , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Linfocitos T Reguladores/inmunología , Proteína del Gen 3 de Activación de Linfocitos
6.
J Virol ; 94(7)2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-31941783

RESUMEN

Chikungunya virus (CHIKV) is an emerging pathogen capable of causing explosive outbreaks. Prior studies showed that exacerbation in arthritogenic alphavirus-induced pathogenesis is attributed to its interaction with multiple immune components, including the complement system. Viremia concomitant to CHIKV infection makes exposure of the virus to complement unavoidable, yet very little is known about CHIKV-complement interactions. Here, we show that CHIKV activated serum complement to modest levels in a concentration- and time-dependent manner, but the virus effectively resisted complement-mediated neutralization. Heat-inactivated serum from seropositive donors could actively neutralize CHIKV due to the presence of potent anti-CHIKV antibodies. Deposition of key complement components C3 and C4 did not alter the resistance of CHIKV to complement. Further, we identified a factor I-like activity in CHIKV that limited complement by inactivating C3b into inactive C3b (iC3b), the complement component known to significantly contribute to disease severity in vivo, but this activity had no effect on C4b. Inactivation of C3b by CHIKV was largely dependent on the concentration of the soluble host cofactor factor H and the virus concentration. A factor I function-blocking antibody had only a negligible effect on the factor I-like activity associated with CHIKV, suggesting that this activity is independent of host factor I and could be of viral origin. Thus, our findings suggest a complement modulatory action of CHIKV which not only helps the virus to evade human complement but may also have implications in alphavirus-induced arthritogenic symptoms.IMPORTANCE Chikungunya virus is a vector-borne pathogen of global significance. The morbidity associated with chikungunya virus (CHIKV) infection, neurovirulence and adaptability to Aedes albopictus, necessitates a deeper understanding of the interaction of CHIKV with the host immune system. Here, we demonstrate that CHIKV is resistant to neutralization by one of the potent barriers of the innate immune arm, the complement system. Chikungunya virus showed marked resistance to complement despite activation and deposition of complement proteins. Interestingly the C3 component associated with the virion was found to be inactive C3b (iC3b), a key factor implicated in the pathogenesis and disease severity in the mouse model of Ross River virus infection. CHIKV also had an associated unique factor I-like activity that mediated the inactivation of C3b into iC3b. We have unraveled a smart strategy adopted by CHIKV to limit complement which has serious implications in viral dissemination, pathogenesis, and disease.


Asunto(s)
Fiebre Chikungunya/inmunología , Activación de Complemento , Complemento C3b/inmunología , Fibrinógeno/inmunología , Adulto , Animales , Anticuerpos Antivirales/inmunología , Virus Chikungunya , Chlorocebus aethiops , Complemento C4/inmunología , Factor H de Complemento/inmunología , Brotes de Enfermedades , Humanos , Pruebas de Neutralización , Células Vero , Replicación Viral
7.
Ann Rheum Dis ; 80(2): 176-184, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32998865

RESUMEN

OBJECTIVES: Autoreactive B cells play a crucial role in the pathogenesis of rheumatoid arthritis (RA), and B cell-depleting therapies using an antibodies, such as rituximab, have been suggested to be effective in RA treatment. However, transient B cell depletion with rituximab is associated with significant safety challenges related to global suppression of the immune system and thus increases the risks of infection and cancer development. To address selective and persistent issues associated with RA therapy, we developed a customised therapeutic strategy employing universal antifluorescein isothiocyanate (FITC) chimeric antigen receptor T cells (CAR-T cells) combined with FITC-labelled antigenic peptide epitopes to eliminate autoreactive B cell subsets recognising these antigens in RA. METHODS: For a proof-of-concept study, four citrullinated peptide epitopes derived from citrullinated autoantigens, namely, citrullinated vimentin, citrullinated type II collagen, citrullinated fibrinogen and tenascin-C, and a cyclocitrulline peptide-1 were selected as ligands for targeting autoreactive B cells; Engineered T cells expressing a fixed anti-FITC CAR were constructed and applied as a universal CAR-T cell system to specifically eliminate these protein-specific autoreactive B cells via recognition of the aforementioned FITC-labelled autoantigenic peptide epitopes. RESULTS: We demonstrated that anti-FITC CAR-T cells could be specifically redirected and kill hybridoma cells generated by immunisation with antigenic peptides, and autoreactive B cell subsets from RA patients via recognition of corresponding FITC-labelled citrullinated peptide epitopes. Additionally, the cytotoxicity of the CAR-T cells was dependent on the presence of the peptides and occurred in a dose-dependent manner. CONCLUSIONS: The approach described here provides a direction for precise, customised approaches to treat RA and can likely be applied to other systemic autoimmune diseases.


Asunto(s)
Artritis Reumatoide/terapia , Fluoresceína-5-Isotiocianato/uso terapéutico , Inmunoterapia Adoptiva/métodos , Receptores de Antígenos de Linfocitos T/uso terapéutico , Receptores Quiméricos de Antígenos/uso terapéutico , Adulto , Células Presentadoras de Antígenos/inmunología , Artritis Reumatoide/inmunología , Autoantígenos/inmunología , Linfocitos B/inmunología , Colágeno Tipo II/inmunología , Epítopos/inmunología , Femenino , Fibrinógeno/inmunología , Humanos , Ligandos , Masculino , Péptidos Cíclicos/inmunología , Prueba de Estudio Conceptual , Tenascina/inmunología , Vimentina/inmunología
8.
PLoS Pathog ; 15(6): e1007816, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31216354

RESUMEN

Fibrinogen is an essential part of the blood coagulation cascade and a major component of the extracellular matrix in mammals. The interface between fibrinogen and bacterial pathogens is an important determinant of the outcome of infection. Here, we demonstrate that a canine host-restricted skin pathogen, Staphylococcus pseudintermedius, produces a cell wall-associated protein (SpsL) that has evolved the capacity for high strength binding to canine fibrinogen, with reduced binding to fibrinogen of other mammalian species including humans. Binding occurs via the surface-expressed N2N3 subdomains, of the SpsL A-domain, to multiple sites in the fibrinogen α-chain C-domain by a mechanism analogous to the classical dock, lock, and latch binding model. Host-specific binding is dependent on a tandem repeat region of the fibrinogen α-chain, a region highly divergent between mammals. Of note, we discovered that the tandem repeat region is also polymorphic in different canine breeds suggesting a potential influence on canine host susceptibility to S. pseudintermedius infection. Importantly, the strong host-specific fibrinogen-binding interaction of SpsL to canine fibrinogen is essential for bacterial aggregation and biofilm formation, and promotes resistance to neutrophil phagocytosis, suggesting a key role for the interaction during pathogenesis. Taken together, we have dissected a bacterial surface protein-ligand interaction resulting from the co-evolution of host and pathogen that promotes host-specific innate immune evasion and may contribute to its host-restricted ecology.


Asunto(s)
Proteínas Bacterianas/inmunología , Biopelículas/crecimiento & desarrollo , Fibrinógeno/inmunología , Evasión Inmune , Inmunidad Innata , Staphylococcus/fisiología , Animales , Proteínas Bacterianas/genética , Pollos , Perros , Fibrinógeno/genética , Humanos
9.
J Autoimmun ; 116: 102542, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32928608

RESUMEN

PURPOSE: Rheumatoid arthritis (RA) is associated with HLA-DRB1 genes encoding a five amino acid basic motive, the shared epitope SE). Each HLA-DRB1 genotype defines a genotype specific risk of developing RA. RA is preceded by the emergence of anti citrullinated protein antibodies (ACPAs). Citrullin is a neutral version of arginin, a basic amino acid, formed after post translational modification by Peptidyl Arginyl Deiminases (PADs). HLA-DRB1 genes associated with RA are also associated with ACPAs. Two models might explain this association. Here we tested both models for prediction of HLA-DRB1 genotypic risks of developing RA. METHODS: We calculated the likelihoods for the 2 HLA-DR molecules encoded by 12 common HLA-DRB1 genotypes to bind at least one randomly chosen peptide from PAD4 or fibrinogen(native or citrullinatd) and compared them with the 12 respective HLA-DRB1genotypic risks of developing RA. RESULTS: HLA-DRB1 Genotypic risks of developing RA correlate with likelihoods of binding PAD4 peptides, not citrullinated Fibrinogen peptides. Thus, the molecular basis for the association of HLA-DR and ACPA positive RA is most likely the capability for RA associated HLA-DR molecules to bind peptides(s) from PAD4.


Asunto(s)
Artritis Reumatoide/inmunología , Cadenas HLA-DRB1/inmunología , Péptidos Cíclicos/inmunología , Péptidos/inmunología , Arginina Deiminasa Proteína-Tipo 4/inmunología , Alelos , Secuencia de Aminoácidos , Artritis Reumatoide/genética , Autoanticuerpos/inmunología , Autoanticuerpos/metabolismo , Autoantígenos/inmunología , Autoantígenos/metabolismo , Unión Competitiva , Citrulinación/inmunología , Epítopos/inmunología , Epítopos/metabolismo , Fibrinógeno/química , Fibrinógeno/inmunología , Fibrinógeno/metabolismo , Predisposición Genética a la Enfermedad/genética , Genotipo , Cadenas HLA-DRB1/genética , Cadenas HLA-DRB1/metabolismo , Humanos , Péptidos/metabolismo , Péptidos Cíclicos/metabolismo , Unión Proteica , Arginina Deiminasa Proteína-Tipo 4/química , Arginina Deiminasa Proteína-Tipo 4/metabolismo
10.
Int Immunol ; 32(8): 533-545, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32239143

RESUMEN

Interstitial lung disease (ILD) is a very common and lethal complication of rheumatoid arthritis (RA), yet its pathogenesis is not well understood, in part due to the lack of adequate animal models. Although collagen-induced arthritis (CIA) is the most widely used animal model for RA, the lung involvement occurring in this model has scarcely been studied. To evaluate the suitability of CIA as a model for RA-associated ILD (RA-ILD), we immunized DBA/1 mice with bovine type II collagen and characterized lung disease in this model. Histologic analyses revealed patchy interstitial infiltration of inflammatory cells in the peripheral regions of the lung, notably in the subpleural region, in mice with CIA. This pattern resembled usual interstitial pneumonia in humans, which is the most prevalent pattern in RA-ILD. Among infiltrates in the lung, CD11bhi macrophages of the M2 phenotype were most prominently increased. IgG and C3 were deposited in the subpleural region where inflammatory cells infiltrated. The sera from CIA mice contained auto-antibodies against citrullinated proteins, which are specific and predictive markers for RA. Protein citrullination was enhanced in the lung of CIA mice compared with naive mice, and citrullinated fibrinogen was primarily targeted by these auto-antibodies. The elevation of auto-antibodies against citrullinated proteins and their deposition in the lung with patchy subpleural preponderance suggest that CIA can serve as a model to study the pathogenesis of RA-ILD.


Asunto(s)
Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Autoanticuerpos/inmunología , Fibrinógeno/inmunología , Enfermedades Pulmonares Intersticiales/inmunología , Animales , Artritis Experimental/inducido químicamente , Artritis Reumatoide/inducido químicamente , Colágeno , Enfermedades Pulmonares Intersticiales/inducido químicamente , Masculino , Ratones , Ratones Endogámicos DBA
11.
Transfusion ; 61(1): 29-34, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33037661

RESUMEN

BACKGROUND: Transfusion medicine standards in Canada state that adult recipients can be transfused with cryoprecipitate of any ABO group, however, not all hospitals follow this guideline. There is a paucity of data on cryoprecipitate anti-A/B levels to reinforce standards. STUDY DESIGN AND METHODS: Manual tube antibody titration was performed on 7 units of group O plasma and the corresponding cryosupernatant plasma and cryoprecipitate. IgG/IgM levels were determined by nephelometry. Additionally, 10 cryoprecipitate each from groups A, B, and O were similarly assessed. From the antibody titer distribution among these samples, the probability of making a pool of cryoprecipitate with a titer ≥1:100 was calculated using bootstrap analysis. RESULTS: Anti-A/B titers in cryoprecipitate were equivalent to those in corresponding plasma; partitioning of anti-A/B activity into cryoprecipitate was not observed. Average IgM concentration was higher in cryoprecipitate than in plasma (P < .01). However, no correlation between IgM levels and anti-A/B titers was established. Among 30 cryoprecipitates from routine blood bank inventory, the median antibody titer and mode were 1:32 and 1:16, respectively. Of the samples tested, 4 of 30 and 9 of 30 had titers above 1:100 and 1:50, respectively. The probability of transfusing an adult dose of cryoprecipitate (pool of 10 cryoprecipitate) with a titer higher than 1:100 was calculated to be less than 1 in 3 million. CONCLUSIONS: This study provides strong evidence to support current Canadian transfusion medicine standards on the safety of transfusion of cryoprecipitate without the need for blood group matching in adult recipients.


Asunto(s)
Sistema del Grupo Sanguíneo ABO/inmunología , Incompatibilidad de Grupos Sanguíneos/inmunología , Transfusión Sanguínea/normas , Factor VIII/inmunología , Fibrinógeno/inmunología , Adulto , Canadá , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Pruebas Inmunológicas , Medición de Riesgo
12.
Transfusion ; 61(3): 660-664, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33491189

RESUMEN

Due to the patients' underlying illness, in combination with circuit-induced coagulopathy, as well as PLT dysfunction, children supported by ECMO are a risk of receiving large volumes of blood components. Given the increasing use of modified blood products and newer biologics, it is unknown whether these products have equal efficacy and safety, in ECMO. The majority of guidance for transfusion therapy is based on expert opinion alone, and research on indications for RBC, plasma, and PLT transfusions for children on ECMO should be a priority.


Asunto(s)
Transfusión Sanguínea/métodos , Oxigenación por Membrana Extracorpórea , Transfusión de Componentes Sanguíneos/efectos adversos , Transfusión de Componentes Sanguíneos/métodos , Niño , Preescolar , Transfusión de Eritrocitos/efectos adversos , Transfusión de Eritrocitos/métodos , Factor VIII/efectos adversos , Factor VIII/inmunología , Fibrinógeno/efectos adversos , Fibrinógeno/inmunología , Humanos , Lactante , Recién Nacido , Plasma , Transfusión de Plaquetas/efectos adversos , Transfusión de Plaquetas/métodos
13.
Acta Pharmacol Sin ; 42(11): 1921-1929, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33633363

RESUMEN

Immune checkpoint inhibition is an important strategy in cancer therapy. Blockade of CTLA-4 and PD-1/PD-L1 is well developed in clinical practice. In the last few years, LAG-3 has received much interest as an emerging novel target in immunotherapy. It was recently reported that FGL1 is a major ligand of LAG-3, which is normally secreted by the liver but is upregulated in several human cancers. FGL1 is a crucial biomarker and target for cancer immunotherapy. As the efficacy of immunotherapy is limited to specific types of patients, the subset of patients needs to be selected appropriately to receive precise treatment according to different biomarkers. To date, there is no test to accurately assess FGL1 expression levels. Nanobodies have some outstanding features, such as high stability, solubility and affinity for diagnostic and therapeutic applications. Here, we report the development and validation of a rapid, sensitive, and cost-effective nanobody-based immunoassay for the detection of FGL1 in human serum. In this study, human FGL1 recombinant protein was expressed and purified for the first time as an immunized antigen. Then, we constructed a nanobody phage display library and screened several nanobodies that bind FGL1 with high affinity. We selected two nanobodies targeting different epitopes of FGL1, one as a capture and the other conjugated with HRP as a probe. The double nanobody-based sandwich ELISA to detect the concentration of FGL1 showed a good response relationship in the range of 15.625-2000 ng/mL, and the recoveries from the spiked sample were in the range of 78% and 100%. This assay could be used as a potential approach for evaluating FGL1 expression for patient stratification and for predicting the therapeutic efficacy of targeting the LAG3/FGL1 axis.


Asunto(s)
Fibrinógeno/inmunología , Fibrinógeno/metabolismo , Anticuerpos de Dominio Único/inmunología , Anticuerpos de Dominio Único/metabolismo , Animales , Camelus , Ensayo de Inmunoadsorción Enzimática/métodos , Células HEK293 , Humanos , Inmunoensayo/métodos
14.
Scand J Immunol ; 92(4): e12925, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32614476

RESUMEN

Fungi are ubiquitous eukaryotic micro-organisms present in virtually all environmental habitats. Although rarely pathogenic to the healthy population, many fungal species are capable of causing human disease in immunocompromised individuals. Thus, fungal infections remain a significant cause of morbidity and mortality, with rising prevalence accompanying the worldwide increase in immunosuppression-based therapies. Therefore, better understanding of the mutual interactions between the protective host mechanisms and the invading fungi remains of critical importance. The innate immune system constitutes the first line of defence against exogenous insults. The innate antifungal immunity is mediated through recognition of specific pathogen-associated molecular patterns (PAMPs) by a broad panel of host pattern recognition receptors (PRRs), responsible for mounting adequate protective responses. In this review, we describe fungal PAMPs as well as a selection of PRRs able to recognize them. We focus on the members of the fibrinogen-related domain (FReD) protein family that have been shown to recognize fungi-derived molecules: ficolins, fibrinogen C domain containing 1 (FIBCD1) and tenascin-C. We describe their structure, their binding targets and their established as well as putative biological functions related to fungal recognition and immunity.


Asunto(s)
Proteínas Fúngicas/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata/inmunología , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Receptores de Reconocimiento de Patrones/inmunología , Animales , Fibrinógeno/inmunología , Humanos , Micosis/inmunología
15.
Clin Lab ; 66(5)2020 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-32390379

RESUMEN

BACKGROUND: This study aimed to evaluate the overall diagnostic value of citrullinated or carbamylated fibrinogen antibodies in patients with rheumatoid arthritis (RA). METHODS: Serum samples collected from 114 patients with established RA, 143 patients with non-RA diseases, and 200 healthy controls were tested by ELISA for citrullinated fibrinogen (Cit-fib), carbamylated fibrinogen (Ca-fib), and chimeric fibrinogen a/b chain citrullinated peptides (CFABCP). Diagnostic indexes and correlations with titers were calculated, cross reactivities of Cit-fib, Ca-fib, and CFABCP were assessed by competition experiments. RESULTS: With a cutoff ensuring 98% specificity for RA patients versus healthy controls, the sensitivities of Cit-fib and Ca-fib are 66.67% and 24.6%, respectively, while the sensitivity of CFABCP was 74.56%. Cit-fib, Ca-fib, and CFABCP can inhibit reciprocally in competition experiments. As for non-RA patients, the positive rate of Ca-fib was higher than that of Cit-fib and CFABCP. CONCLUSIONS: Citrullination and carbamylation of fibrinogen both have a role in RA diagnosing, but citrullination is better. The recombination of peptides, CFABCP, has high specificity and considerable sensitivity for diagnosis for RA patients.


Asunto(s)
Artritis Reumatoide/diagnóstico , Autoanticuerpos/sangre , Autoantígenos/inmunología , Fibrinógeno/inmunología , Anciano , Autoantígenos/química , Citrulinación , Ensayo de Inmunoadsorción Enzimática , Femenino , Fibrinógeno/química , Humanos , Masculino , Persona de Mediana Edad , Péptidos/química , Péptidos/inmunología , Carbamilación de Proteína , Sensibilidad y Especificidad
16.
Rheumatol Int ; 40(11): 1873-1881, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32472302

RESUMEN

Treatment recommendations of early rheumatoid arthritis (RA) suggest differential management of patients on the basis of prognostic factors. In this study we aimed to investigate the relationship between autoantibodies against a novel citrullinated fibrinogen peptide (anti-CFP), smoking status, clinical activity and therapeutic response in Cuban patients with early RA, receiving treatment with methotrexate in comparison to rheumatoid factor (RF), anti-cyclic citrullinated peptide of second generation (anti-CCP2) and anti-mutated citrullinated vimentin (anti-MCV). A 6-month prospective observational study was performed in 60 early RA patients at baseline and 6 months after receiving methotrexate. Baseline and outcome measures included disease activity score of 28 joints (DAS 28), simplified disease activity index (SDAI), anti-CFP antibodies, RF, anti-CCP2 and anti-MCV. Therapeutic response was determined using 20/50/70 American College of Rheumatology (ACR) response rates. DAS28 (p < 0.0001), SDAI (p < 0.0001) as well as titres of anti-CFP (p = 0.0481), anti-CCP2 (p = 0.0082), RF IgM (p = 0.0187) and RF IgA (p = 0.0252) decreased under therapy. Multivariate analyses showed association of final anti-CFP values with sex and smoking status (p = 0.0296). It is of note that anti-CFP antibodies were one of predictors for DAS 28 (p = 0.0072) SDAI (p < 0.0001) and ACR response (p = 0.0003) in multivariate models. Anti-CFP antibodies decrease in correspondence with clinical improvement after 6-month therapy and are associated with sex and smoking status. Moreover, baseline anti-CFP antibodies, using in combination with sex, smoking status and autoantibodies (anti-CCP2, anti-MCV or RF) seems to have clinical relevance for predicting clinical activity and therapeutic response.


Asunto(s)
Anticuerpos Antiproteína Citrulinada/inmunología , Artritis Reumatoide/inmunología , Fibrinógeno/inmunología , Fumar/inmunología , Adulto , Antirreumáticos/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/epidemiología , Artritis Reumatoide/fisiopatología , Cuba , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Masculino , Metotrexato/uso terapéutico , Persona de Mediana Edad , Índice de Severidad de la Enfermedad , Fumar/epidemiología , Resultado del Tratamiento
17.
Proc Natl Acad Sci U S A ; 114(47): E10169-E10177, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29109281

RESUMEN

Autoantibodies to citrullinated proteins (ACPAs) are present in two-thirds of patients with rheumatoid arthritis (RA). ACPAs are produced in the absence of identified T cell responses for each citrullinated protein. Peptidyl arginine deiminase 4 (PAD4), which binds proteins and citrullinates them, is the target of autoantibodies in early RA. This suggests a model for the emergence of ACPAs in the absence of detectable T cells specific for citrullinated antigens: ACPAs could arise because PADs are recognized by T cells, which help the production of autoantibodies to proteins bound by PADs, according to a "hapten/carrier" model. Here, we tested this model in normal mice. C3H are healthy mice whose IEßk chain is highly homologous to the ß1 chain HLA-DRB1*04:01, the allele most strongly associated with RA in humans. C3H mice immunized with PADs developed antibodies and T cells to PAD and IgG antibodies to citrullinated fibrinogen peptides, in the absence of a T cell response to fibrinogen. To analyze the MHC background effect on hapten/carrier immunization, we immunized DBA/2 mice (whose IEßd chain is similar to that of HLA-DRB1*04:02, an HLA-DR4 subtype not associated with RA). DBA/2 mice failed to develop antibodies to citrullinated fibrinogen peptides. Thus, T cell immunization to PAD proteins may trigger ACPAs through a hapten/carrier mechanism. This may constitute the basis for a new mouse model of ACPA-positive RA.


Asunto(s)
Artritis Reumatoide/inmunología , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Citrulinación/inmunología , Desiminasas de la Arginina Proteica/inmunología , Animales , Citrulina/metabolismo , Modelos Animales de Enfermedad , Fibrinógeno/inmunología , Fibrinógeno/metabolismo , Haptenos/inmunología , Humanos , Inmunización/métodos , Complejo Mayor de Histocompatibilidad/inmunología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos DBA , Linfocitos T/inmunología
18.
J Biol Chem ; 293(35): 13578-13591, 2018 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-30002122

RESUMEN

Some strains of the bacterial pathogen Streptococcus pyogenes secrete protein SIC (streptococcal inhibitor of complement), including strains of the clinically relevant M1 serotype. SIC neutralizes the effect of a number of antimicrobial proteins/peptides and interferes with the function of the host complement system. Previous studies have shown that some S. pyogenes proteins bind and modulate coagulation and fibrinolysis factors, raising the possibility that SIC also may interfere with the activity of these factors. Here we show that SIC interacts with both human thrombin and plasminogen, key components of coagulation and fibrinolysis. We found that during clot formation, SIC binds fibrin through its central region and that SIC inhibits fibrinolysis by interacting with plasminogen. Flow cytometry results indicated that SIC and plasminogen bind simultaneously to S. pyogenes bacteria, and fluorescence microscopy revealed co-localization of the two proteins at the bacterial surface. As a consequence, SIC-expressing bacteria entrapped in clots inhibit fibrinolysis, leading to delayed bacterial escape from the clots as compared with mutant bacteria lacking SIC. Moreover, within the clots SIC-expressing bacteria were protected against killing. In an animal model of subcutaneous infection, SIC-expressing bacteria exhibited a delayed systemic spread. These results demonstrate that the bacterial protein SIC interferes with coagulation and fibrinolysis and thereby enhances bacterial survival, a finding that has significant implications for S. pyogenes virulence.


Asunto(s)
Proteínas Bacterianas/inmunología , Fibrinólisis , Infecciones Estreptocócicas/inmunología , Streptococcus pyogenes/inmunología , Trombosis/inmunología , Animales , Proteínas del Sistema Complemento/inmunología , Femenino , Fibrina/inmunología , Fibrinógeno/inmunología , Humanos , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Infecciones Estreptocócicas/complicaciones , Infecciones Estreptocócicas/microbiología , Trombina/inmunología , Trombosis/complicaciones , Trombosis/microbiología
19.
Ann Rheum Dis ; 78(2): 186-191, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30552174

RESUMEN

OBJECTIVES: Controlled immune responses rely on integrated crosstalk between cells and their microenvironment. We investigated whether targeting proinflammatory signals from the extracellular matrix that persist during pathological inflammation provides a viable strategy to treat rheumatoid arthritis (RA). METHODS: Monoclonal antibodies recognising the fibrinogen-like globe (FBG) of tenascin-C were generated by phage display. Clones that neutralised FBG activation of toll-like receptor 4 (TLR4), without impacting pathogenic TLR4 activation, were epitope mapped by crystallography. Antibodies stained synovial biopsies of patients at different stages of RA development. Antibody efficacy in preventing RA synovial cell cytokine release, and in modulating collagen-induced arthritis in rats, was assessed. RESULTS: Tenascin-C is expressed early in the development of RA, even before disease diagnosis, with higher levels in the joints of people with synovitis who eventually developed RA than in people whose synovitis spontaneously resolved. Anti-FBG antibodies inhibited cytokine release by RA synovial cells and prevented disease progression and tissue destruction during collagen-induced arthritis. CONCLUSIONS: Early changes in the synovial microenvironment contribute to RA progression; blocking proinflammatory signals from the matrix can ameliorate experimental arthritis. These data highlight a new drug class that could offer early, disease-specific immune modulation in RA, without engendering global immune suppression.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Artritis Reumatoide/inmunología , Microambiente Celular/inmunología , Inmunoterapia/métodos , Membrana Sinovial/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Artritis Experimental , Colágeno , Citocinas/metabolismo , Progresión de la Enfermedad , Fibrinógeno/inmunología , Humanos , Ratas , Tenascina/metabolismo , Receptor Toll-Like 4/inmunología
20.
J Allergy Clin Immunol ; 141(4): 1439-1449, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28807602

RESUMEN

BACKGROUND: The pathogenesis of macrophage activation syndrome (MAS) is not clearly understood: a large body of evidence supports the involvement of mechanisms similar to those implicated in the setting of primary hemophagocytic lymphohistiocytosis. OBJECTIVE: We sought to investigate the pathogenic role of IFN-γ and the therapeutic efficacy of IFN-γ neutralization in an animal model of MAS. METHODS: We used an MAS model established in mice transgenic for human IL-6 (IL-6TG mice) challenged with LPS (MAS mice). Levels of IFN-γ and IFN-γ-inducible chemokines were evaluated by using real-time PCR in the liver and spleen and by means of ELISA in plasma. IFN-γ neutralization was achieved by using the anti-IFN-γ antibody XMG1.2 in vivo. RESULTS: Mice with MAS showed a significant upregulation of the IFN-γ pathway, as demonstrated by increased mRNA levels of Ifng and higher levels of phospho-signal transducer and activator of transcription 1 in the liver and spleen and increased expression of the IFN-γ-inducible chemokines Cxcl9 and Cxcl10 in the liver and spleen, as well as in plasma. A marked increase in Il12a and Il12b expression was also found in livers and spleens of mice with MAS. In addition, mice with MAS had a significant increase in numbers of liver CD68+ macrophages. Mice with MAS treated with an anti-IFN-γ antibody showed a significant improvement in survival and body weight recovery associated with a significant amelioration of ferritin, fibrinogen, and alanine aminotransferase levels. In mice with MAS, treatment with the anti-IFN-γ antibody significantly decreased circulating levels of CXCL9, CXCL10, and downstream proinflammatory cytokines. The decrease in CXCL9 and CXCL10 levels paralleled the decrease in serum levels of proinflammatory cytokines and ferritin. CONCLUSION: These results provide evidence for a pathogenic role of IFN-γ in the setting of MAS.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Interferón gamma/inmunología , Síndrome de Activación Macrofágica/inmunología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Alanina Transaminasa/inmunología , Animales , Quimiocina CXCL10/inmunología , Quimiocina CXCL9/inmunología , Citocinas/inmunología , Modelos Animales de Enfermedad , Ferritinas/inmunología , Fibrinógeno/inmunología , Inflamación/inmunología , Linfohistiocitosis Hemofagocítica/inmunología , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA